Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Genes Dev ; 36(15-16): 887-900, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-36167470

RESUMO

The polycomb complex component Bmi1 promotes the maintenance of stem cells in multiple postnatal tissues, partly by negatively regulating the expression of p16Ink4a and p19Arf, tumor suppressors associated with cellular senescence. However, deficiency for p16Ink4a and p19Arf only partially rescues the function of Bmi1-deficient stem cells. We conditionally deleted Bmi1 from adult hematopoietic cells and found that this slowly depleted hematopoietic stem cells (HSCs). Rather than inducing senescence, Bmi1 deficiency increased HSC division. The increased cell division was caused partly by increased Aristaless-related homeobox (ARX) transcription factor expression, which also increased ribosomal RNA expression. However, ARX deficiency did not rescue HSC depletion. Bmi1 deficiency also increased protein synthesis, protein aggregation, and protein ubiquitylation independent of its effects on cell division and p16Ink4a, p19Arf, and ARX expression. Bmi1 thus promotes HSC quiescence by negatively regulating ARX expression and promotes proteostasis by suppressing protein synthesis. This highlights a new connection between the regulation of stem cell maintenance and proteostasis.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina , Proteostase , Inibidor p16 de Quinase Dependente de Ciclina/genética , Células-Tronco Hematopoéticas , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Agregados Proteicos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Ribossômico/metabolismo
2.
Nat Cell Biol ; 24(5): 697-707, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35513711

RESUMO

How are haematopoietic stem cells (HSCs) protected from inflammation, which increases with age and can deplete HSCs? Adiponectin, an anti-inflammatory factor that is not required for HSC function or haematopoiesis, promotes stem/progenitor cell proliferation after bacterial infection and myeloablation. Adiponectin binds two receptors, AdipoR1 and AdipoR2, which have ceramidase activity that increases upon adiponectin binding. Here we found that adiponectin receptors are non-cell-autonomously required in haematopoietic cells to promote HSC quiescence and self-renewal. Adiponectin receptor signalling suppresses inflammatory cytokine expression by myeloid cells and T cells, including interferon-γ and tumour necrosis factor. Without adiponectin receptors, the levels of these factors increase, chronically activating HSCs, reducing their self-renewal potential and depleting them during ageing. Pathogen infection accelerates this loss of HSC self-renewal potential. Blocking interferon-γ or tumour necrosis factor signalling partially rescues these effects. Adiponectin receptors are thus required in immune cells to sustain HSC quiescence and to prevent premature HSC depletion by reducing inflammation.


Assuntos
Adiponectina , Receptores de Adiponectina , Adiponectina/genética , Adiponectina/metabolismo , Adulto , Células-Tronco Hematopoéticas/metabolismo , Humanos , Inflamação/metabolismo , Interferon gama/genética , Interferon gama/metabolismo , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo , Fatores de Necrose Tumoral/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(27): 13311-13319, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31209047

RESUMO

Cellular senescence defines an irreversible cell growth arrest state linked to loss of tissue function and aging in mammals. This transition from proliferation to senescence is typically characterized by increased expression of the cell-cycle inhibitor p16INK4a and formation of senescence-associated heterochromatin foci (SAHF). SAHF formation depends on HIRA-mediated nucleosome assembly of histone H3.3, which is regulated by the serine/threonine protein kinase Pak2. However, it is unknown if Pak2 contributes to cellular senescence. Here, we show that depletion of Pak2 delayed oncogene-induced senescence in IMR90 human fibroblasts and oxidative stress-induced senescence of mouse embryonic fibroblasts (MEFs), whereas overexpression of Pak2 accelerated senescence of IMR90 cells. Importantly, depletion of Pak2 in BubR1 progeroid mice attenuated the onset of aging-associated phenotypes and extended life span. Pak2 is required for expression of genes involved in cellular senescence and regulated the deposition of newly synthesized H3.3 onto chromatin in senescent cells. Together, our results demonstrate that Pak2 is an important regulator of cellular senescence and organismal aging, in part through the regulation of gene expression and H3.3 nucleosome assembly.


Assuntos
Envelhecimento , Senescência Celular , Quinases Ativadas por p21/fisiologia , Envelhecimento/metabolismo , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica , Histonas/metabolismo , Longevidade , Camundongos Knockout , Estresse Oxidativo , Reação em Cadeia da Polimerase em Tempo Real , Quinases Ativadas por p21/metabolismo
4.
Nature ; 549(7673): 476-481, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28825709

RESUMO

Stem-cell fate can be influenced by metabolite levels in culture, but it is not known whether physiological variations in metabolite levels in normal tissues regulate stem-cell function in vivo. Here we describe a metabolomics method for the analysis of rare cell populations isolated directly from tissues and use it to compare mouse haematopoietic stem cells (HSCs) to restricted haematopoietic progenitors. Each haematopoietic cell type had a distinct metabolic signature. Human and mouse HSCs had unusually high levels of ascorbate, which decreased with differentiation. Systemic ascorbate depletion in mice increased HSC frequency and function, in part by reducing the function of Tet2, a dioxygenase tumour suppressor. Ascorbate depletion cooperated with Flt3 internal tandem duplication (Flt3ITD) leukaemic mutations to accelerate leukaemogenesis, through cell-autonomous and possibly non-cell-autonomous mechanisms, in a manner that was reversed by dietary ascorbate. Ascorbate acted cell-autonomously to negatively regulate HSC function and myelopoiesis through Tet2-dependent and Tet2-independent mechanisms. Ascorbate therefore accumulates within HSCs to promote Tet activity in vivo, limiting HSC frequency and suppressing leukaemogenesis.


Assuntos
Ácido Ascórbico/metabolismo , Carcinogênese/metabolismo , Células-Tronco Hematopoéticas/citologia , Leucemia/patologia , Animais , Ácido Ascórbico/análise , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/metabolismo , Carcinogênese/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia/genética , Masculino , Metabolômica , Camundongos , Mielopoese/genética , Proteínas Proto-Oncogênicas/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
5.
Elife ; 3: e02669, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24843006

RESUMO

Neurosphere formation is commonly used as a surrogate for neural stem cell (NSC) function but the relationship between neurosphere-initiating cells (NICs) and NSCs remains unclear. We prospectively identified, and isolated by flow cytometry, adult mouse lateral ventricle subventricular zone (SVZ) NICs as Glast(mid)EGFR(high)PlexinB2(high)CD24(-/low)O4/PSA-NCAM(-/low)Ter119/CD45(-) (GEPCOT) cells. They were highly mitotic and short-lived in vivo based on fate-mapping with Ascl1(CreERT2) and Dlx1(CreERT2). In contrast, pre-GEPCOT cells were quiescent, expressed higher Glast, and lower EGFR and PlexinB2. Pre-GEPCOT cells could not form neurospheres but expressed the stem cell markers Slc1a3-CreER(T), GFAP-CreER(T2), Sox2(CreERT2), and Gli1(CreERT2) and were long-lived in vivo. While GEPCOT NICs were ablated by temozolomide, pre-GEPCOT cells survived and repopulated the SVZ. Conditional deletion of the Bmi-1 polycomb protein depleted pre-GEPCOT and GEPCOT cells, though pre-GEPCOT cells were more dependent upon Bmi-1 for Cdkn2a (p16(Ink4a)) repression. Our data distinguish quiescent NSCs from NICs and make it possible to study their properties in vivo.DOI: http://dx.doi.org/10.7554/eLife.02669.001.


Assuntos
Envelhecimento/metabolismo , Células-Tronco Neurais/citologia , Prosencéfalo/citologia , Esferoides Celulares/citologia , Animais , Antimitóticos/farmacologia , Proliferação de Células , Separação Celular , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Integrases/metabolismo , Camundongos Endogâmicos C57BL , Neurogênese/efeitos dos fármacos , Neuroglia/citologia , Neuroglia/metabolismo , Fenótipo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Temozolomida
6.
Nat Struct Mol Biol ; 20(1): 14-22, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23288364

RESUMO

Nucleosome assembly following DNA replication, DNA repair and gene transcription is critical for the maintenance of genome stability and epigenetic information. Nucleosomes are assembled by replication-coupled or replication-independent pathways with the aid of histone chaperone proteins. How these different nucleosome assembly pathways are regulated remains relatively unclear. Recent studies have provided insight into the mechanisms and the roles of histone chaperones in regulating nucleosome assembly. Alterations or mutations in factors involved in nucleosome assembly have also been implicated in cancer and other human diseases. This review highlights the recent progress and outlines future challenges in the field.


Assuntos
Replicação do DNA , Chaperonas de Histonas/metabolismo , Neoplasias/metabolismo , Nucleossomos/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Montagem e Desmontagem da Cromatina , Instabilidade Genômica , Histonas/genética , Histonas/metabolismo , Humanos , Neoplasias/genética
7.
Cell Cycle ; 9(15): 2979-85, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20699646

RESUMO

The process of coordinated DNA replication and nucleosome assembly, termed replication-coupled (RC) nucleosome assembly, is important for the maintenance of genome integrity. Loss of genome integrity is linked to aging and cancer. RC nucleosome assembly involves deposition of histone H3-H4 by the histone chaperones CAF-1, Rtt106 and Asf1 onto newly-replicated DNA. Coordinated actions of these three histone chaperones are regulated by modifications on the histone proteins. One such modification is histone H3 lysine 56 acetylation (H3K56Ac), a mark of newly-synthesized histone H3 that regulates the interaction between H3-H4 and the histone chaperones CAF-1 and Rtt106 following DNA replication and DNA repair. Recently, we have shown that the lysine acetyltransferase Gcn5 and H3 N-terminal tail lysine acetylation also regulates the interaction between H3-H4 and CAF-1 to promote the deposition of newly-synthesized histones. Genetic studies indicate that Gcn5 and Rtt109, the H3K56Ac lysine acetyltransferase, function in parallel to maintain genome stability. Utilizing synthetic genetic array analysis, we set out to identify additional genes that function in parallel with Gcn5 in response to DNA damage. We summarize here the role of Gcn5 in nucleosome assembly and suggest that Gcn5 impacts genome integrity via multiple mechanisms, including nucleosome assembly.


Assuntos
Instabilidade Genômica , Histona Acetiltransferases/metabolismo , Nucleossomos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Acetilação , Animais , Reparo do DNA , Replicação do DNA , Humanos , Modelos Biológicos , Saccharomyces cerevisiae/metabolismo
8.
Protein Cell ; 1(7): 607-12, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21203931

RESUMO

Chromatin structure governs a number of cellular processes including DNA replication, transcription, and DNA repair. During DNA replication, chromatin structure including the basic repeating unit of chromatin, the nucleosome, is temporarily disrupted, and then reformed immediately after the passage of the replication fork. This coordinated process of nucleosome assembly during DNA replication is termed replication-coupled nucleosome assembly. Disruption of this process can lead to genome instability, a hallmark of cancer cells. Therefore, addressing how replication-coupled nucleosome assembly is regulated has been of great interest. Here, we review the current status of this growing field of interest, highlighting recent advances in understanding the regulation of this important process by the dynamic interplay of histone chaperones and histone modifications.


Assuntos
Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Nucleossomos/metabolismo , Acetilação , Animais , Replicação do DNA , Humanos , Conformação de Ácido Nucleico , Processamento de Proteína Pós-Traducional
10.
Pharmacogenet Genomics ; 18(12): 1083-94, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18820593

RESUMO

OBJECTIVE: Thiopurine S-methyltransferase (TPMT)*3A is degraded much more rapidly than is the 'wild-type' enzyme through a ubiquitin-proteasome-dependent process. It also forms aggresomes, suggesting a possible dynamic balance between degradation and aggregation. We set out to identify genes encoding proteins participating in these processes. METHODS: Green fluorescent protein tagged TPMT*3A was expressed in a Saccharomyces cerevisiae gene deletion library, and flow cytometry was used to screen for cells with high fluorescence intensity, indicating the loss of a gene essential for TPMT*3A degradation. RESULTS: Twenty-four yeast genes were identified in functional categories that included ubiquitin-dependent protein degradation, vesicle trafficking, and vacuolar degradation. The presence of genes encoding proteins involved in vesicular transport and vacuolar degradation suggested a possible role in TPMT*3A degradation for autophagy--a process not previously identified as a pharmacogenomic mechanism. In support of that hypothesis, TPMT*3A aggregates increased dramatically in mutants for vacuolar protease and autophagy-related genes. Furthermore, TPMT*3A expression in human cells induced autophagy, and small interfering RNA-mediated knockdown of ATG7, an autophagy-related human protein, enhanced TPMT*3A aggregation but not that of TPMT*3C or wild-type TPMT, indicating that autophagy contributes to TPMT*3A degradation in mammalian cells. We also demonstrated that UBE2G2, the human homologue of the E2 ubiquitin-conjugating enzyme identified during the yeast genetic screen, was involved in TPMT*3A degradation in human cells. CONCLUSION: These results indicate that autophagy should be considered among mechanisms responsible for the effects of pharmacogenetically significant polymorphisms that alter encoded amino acids.


Assuntos
Autofagia , Metiltransferases/genética , Metiltransferases/metabolismo , Processamento de Proteína Pós-Traducional , Saccharomyces cerevisiae/enzimologia , Linhagem Celular , Dineínas/metabolismo , Citometria de Fluxo , Deleção de Genes , Genes Fúngicos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Isoenzimas , Metiltransferases/química , Microtúbulos/metabolismo , Chaperonas Moleculares/metabolismo , Estrutura Quaternária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA